Challenges and Solutions in Cell Therapy Development
Credit: iStock
Cell therapy involves the transplantation of live human cells into a patient to repair damaged tissue or cure disease.
In this article, we take a look at how researchers are tackling some of the challenges that are preventing cell therapies from being more widely used, paying particular attention to cellular immunotherapies for cancer and induced pluripotent stem cells (iPSCs) for regenerative medicine.
Download this article to learn more about:
- Non-stem cell and stem cell-based therapies
- Next-generation cellular immunotherapies
- Speeding up production of stem cell-based therapeutics
Article
1
Challenges and Solutions in Cell
Therapy Development
Monica Hoyos Flight, PhD
Cell therapy involves the transplantation of live human cells into a patient to repair damaged tissue or
cure disease. A variety of cell types either belonging to the patient, in the case of autologous therapies, or
from a donor – known as allogeneic cell therapies – can be used.
Although cell therapies in the form of bone marrow transplants have been used to treat rare forms of
blood cancer since the 1950s, most commercially available cell-based therapies today have been approved within the past decade.
To date, 27 cell therapies have been approved by the United States Food and Drug Administration (FDA)
for the treatment of multiple indications, including various forms of cancer, inherited metabolic disorders
and immune system disorders.1
Many more are likely to be submitted for approval soon, given the over
1,600 ongoing clinical trials for cell therapies registered with ClinicalTrials.gov.2
In this article, we take a look at how researchers are tackling some of the challenges that are preventing cell therapies from being more widely used, paying particular attention to cellular immunotherapies
for cancer and induced pluripotent stem cells (iPSC) for regenerative medicine. We also outline some
advances in manufacturing and regulatory approval processes that will help expand patient access to
innovative, life-changing treatments.
Non-stem cell-based therapies
These types of therapies involve somatic cells from a patient or a donor, such as fibroblasts, pancreatic
islet cells or immune cells. Examples include allogeneic cultured keratinocytes for the treatment of adults
with thermal burns and allogeneic pancreatic islet cells for people with type 1 diabetes who are unable to
maintain healthy blood sugar levels despite thorough diabetes management and education.
The most commonly used somatic cells are T cells, white blood cells that are critical for triggering an
immune response to pathogens, allergens and tumors. The first chimeric antigen receptor (CAR) T cell to
be approved by the FDA in 2017 was invented by Professor Carl June at the University of Pennsylvania.
This CAR T-cell therapy – known as tisagenlecleucel (Kymriah®) – can be used for the treatment of certain
pediatric and young adult patients with advanced chronic lymphocytic leukemia.
Currently, there are six FDA-approved CAR T-cell therapies for the treatment of various lymphomas and
myeloma. These cellular immunotherapies involve collecting T cells from the patient’s blood and mod-
CHALLENGES AND SOLUTIONS IN CELL THERAPY DEVELOPMENT 2
Article
ifying them genetically in the laboratory so that they make CARs that track down cancer-associated
antigens. When re-infused into the patient, the modified T cells attach to and kill the cancer cells, thereby
helping to clear the cancer from the body.
Dr. Neil Sheppard, a director in Prof. Carl June’s laboratory, highlights the advantages of cellular immunotherapies for cancer: “We know it is possible for the immune system to completely cure us of even
advanced diseases, including cancer, under the right circumstances and potentially without damaging
healthy tissues,” he explains.
While he acknowledges that a lot of work remains to be done to realize the full potential of these therapies, he is optimistic about progress. “With various genetic, epigenetic and RNA-editing tools the promise
of cellular immunotherapies is within our grasp,” he says.
Next-generation cellular immunotherapies
Despite the remarkable efficacy of CAR T-cell therapies against B-cell malignancies, patients can experience recurrence if the cancer cells stop expressing the targeted antigen, and CAR T-cell therapies have
also shown limited efficacy against solid tumors.3,4
To address these challenges, Sheppard and others are pursuing several approaches. These include developing CAR T cells that target multiple antigens to prevent immune escape and using alternative cell types
such as natural killer (NK) cells, which do not mediate graft-versus-host disease and might be easier to
use in an allogeneic setting.5
“Solid tumors remain a challenge,” says Sheppard. “To achieve CAR T efficacy in solid tumors, target
antigens such as claudin 6 and claudin 18.2 are being utilized, sometimes in conjunction with vaccination
with the antigen to boost CAR T-cell proliferation and activity.”
There is growing evidence that intratumoral injection of CAR T cells can have a greater effect compared
to intravenous infusion. Moreover, combining CAR T cells with oncolytic viruses that attract CAR T cells to
the tumor aids tumor infiltration and strengthens resistance to the immunosuppressive tumor microenvironment.6
The recent approval of the first tumor-infiltrating lymphocyte (TIL) therapy raises new hope for cell therapies in solid cancers.7
TILs have an innate ability to seek out cancer neoantigens, fragments of mutated
intracellular proteins that are presented on the surface of cancer cells. One such treatment for unresectable or metastatic melanoma – lifileucel (Amtagvi™) – involves the removal of TILs from patients’ tumor
tissue, growing them in bioreactors and re-infusing them into patients to destroy cancer cells expressing
these patient-specific antigens.
Stem cell-based therapies
Stem cell treatments play a key role in regenerative medicine as their ability to self-renew and differentiate into specialized cells can be used to replace damaged or diseased cells and restore normal function.
Dr. Shinya Yamanaka’s groundbreaking discovery that adult cells can be reprogrammed into pluripotent
stem cells through the expression of embryonic transcription factors offers researchers the opportunity
to generate patient-specific regenerative cell therapies without having to rely on controversial embryonic
stem cells or worry about immune rejection.8
CHALLENGES AND SOLUTIONS IN CELL THERAPY DEVELOPMENT 3
Article
Since then, great progress has been made in generating and differentiating iPSCs. “iPSCs are remarkably
useful, and in a class by themselves,” says Professor Jeanne Loring, founding director of the Center for
Regenerative Medicine and emeritus professor at the Scripps Research Institute in La Jolla, California.
“They are the only non-cancer cells that can continue to divide indefinitely in a culture dish, and retain the
ability to give rise to every cell type in the body.”
Loring’s research focuses on developing iPSC-based therapies for Parkinson’s disease, multiple sclerosis
and autism. She believes these therapies hold great promise for conditions known to be caused by the
loss of specific cell types. However, there are still several obstacles associated with iPSC-based therapies
that need to be addressed before they can enter clinical trials.
“The development of iPSC-derived cell therapies requires cutting-edge technologies in multiple areas –
from making the stem cells and differentiating them into the right cell type in culture, to determining the
best stage of differentiation for transplanting the cells and sophisticated genomics and bioinformatics,
with machine learning to make sure the cells are at the right stage and that they don’t contain dangerous
mutations,” Loring explains.
In her opinion, technology is key to overcoming these challenges. “This is a field where all of the work
has been hands-on with highly skilled people, which makes it very expensive,“ she says. “The use of AI
to make decisions about the culture of the cells and new genomics methods that ensure the integrity of
the genomes of cells before they are transplanted, are just a couple of examples of ways to advance cell
therapies to the clinic.”
Manufacturing innovations
In addition to sharing the main challenges of drug development, such as the need to demonstrate safety,
target engagement and pharmacological activity, cell therapies face unique challenges. “The manufacturing process for CAR T-cell therapy greatly affects the potency of cell therapy product,” says Sheppard.
His team is exploring how to scale up their cell manufacturing process in the laboratory into a large-scale
good manufacturing process (GMP) needed for clinical trials or even a commercial scale process designed to serve thousands of patients each year.
So far, they have learned that, with patient-specific immune cells, the longer the duration of the cell manufacturing process and the more manipulation steps involved, the lower the potency of the emerging cell.
“The evidence shows that CAR T-cell products made in manufacturing processes as short as one to three
days have superior activity once inside the human body compared to those made over nine days or more.”
In the not-too-distant future, Sheppard envisages the creation of self-contained GMP machines where a
patient’s T cells are loaded and the chosen type of CAR T-cell therapy is produced at the press of a button
in a hospital setting. “I imagine the future of CAR T-cell therapy production being as simple from the
prescriber’s perspective as using the office coffee machine – load the desired sachet (gene vector), water
(reagents) and a cup (cells), press the button and your work is done.”
Speeding up production of stem cell-based therapeutics
Since 2003, the UK’s Stem Cell Bank (UKSCB) has been supporting the development of pluripotent stem
cell-derived therapies and driving improvements in the manufacturing process to produce quality-controlled pluripotent stem cell lines (iPSCs and human embryonic stem cells, or hESCs) that can be differentiated into specialized cell types for tissue regeneration. At the 20th anniversary of the cell repository
last October, Dr. Lee Carpenter, the head of the UKSCB, highlighted automation and machine learning as
CHALLENGES AND SOLUTIONS IN CELL THERAPY DEVELOPMENT 4
Article
being key to accelerating the safe manufacture of stem cell-derived therapies and giving patients faster
access to potentially life-saving treatments.9
The UKSCB has demonstrated, using a robotic system, that clinical-grade pluripotent stem cell production
is possible and is currently comparing the automated process to manual production. The use of automation, robotics and modular production platforms will not just help reduce reliance on manual operations
and variability in process performance, it will also enable scalability and drive down costs. “There is a
race to develop the instrumentation to automate stem cell-derived cell therapy manufacturing,” says
Loring.
Streamlining the approval process
The final hurdle when developing cell therapies is obtaining regulatory approval. The review of a New
Drug Application (NDA) by the FDA can take up to a year, significantly delaying the commercialization of a
new therapy.10
Cell therapies are evolving quickly, and so too must the regulations. Regulatory agencies are adapting
to the unique challenges of cell therapies by implementing pathways aimed at expediting assessments,
such as the FDA’s Regenerative Medicine Advanced Therapy (RMAT) designation and the European Medicines Agency (EMA)’s Priority Medicines (PRIME) scheme.11 In addition, the FDA is on a recruitment drive
to hire more staff in its Office of Therapeutic Products to tackle the increasing workload.12
Efforts to increase the dialog between regulators, industry and academia to harmonize regulatory standards are also starting to bear fruit. The benefits of international programs to build regulatory capacity
and share information are evidenced by multinational approvals of CAR T-cell therapies.13 As the cell therapy pipeline continues to expand, increasing the alignment of regulatory pathways across countries will
be crucial to facilitating access to new transformative treatments to patients around the world.
Sponsored by
Download the Whitepaper for FREE Now!
Information you provide will be shared with the sponsors for this content.
Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version